研究者業績

横山 真隆

ヨコヤマ マサタカ  (Masataka Yokoyama)

基本情報

所属
千葉大学 大学院医学研究院分子病態解析学 助教
学位
博士(医学)(千葉大学.)

J-GLOBAL ID
201801004970118539
researchmap会員ID
B000306565

外部リンク

千葉大学大学院医学研究院分子病態解析学で、基礎から臨床への橋渡し研究を目指しています。2014年より2019年春まで、Weill Cornell Medical College, Shahin Rafii研にて血管内皮細胞研究に専念したのち、現在は心臓血管内皮細胞の臓器特異的役割の解明と血管内皮細胞のストレス応答反応をテーマに研究しております。

メインテーマである「心臓血管内皮細胞の臓器特異性研究」は、血管における転写因子の調節・発現コンロトールを次世代シークエンス技術を中心に俯瞰するユニークな研究で、将来臨床治療材料となりうる血管内皮細胞の利用を目標としています。

研究キーワード

 3

学歴

 3

委員歴

 1

主要な論文

 103
  • Masanori Fujimoto, Masataka Yokoyama, Masahiro Kiuchi, Hiroyuki Hosokawa, Akitoshi Nakayama, Naoko Hashimoto, Ikki Sakuma, Hidekazu Nagano, Kazuyuki Yamagata, Fujimi Kudo, Ichiro Manabe, Eunyoung Lee, Ryo Hatano, Atsushi Onodera, Kiyoshi Hirahara, Koutaro Yokote, Takashi Miki, Toshinori Nakayama, Tomoaki Tanaka
    Nature communications 13(1) 5408-5408 2022年9月15日  
    The liver stores glycogen and releases glucose into the blood upon increased energy demand. Group 2 innate lymphoid cells (ILC2) in adipose and pancreatic tissues are known for their involvement in glucose homeostasis, but the metabolic contribution of liver ILC2s has not been studied in detail. Here we show that liver ILC2s are directly involved in the regulation of blood glucose levels. Mechanistically, interleukin (IL)-33 treatment induces IL-13 production in liver ILC2s, while directly suppressing gluconeogenesis in a specific Hnf4a/G6pc-high primary hepatocyte cluster via Stat3. These hepatocytes significantly interact with liver ILC2s via IL-13/IL-13 receptor signaling. The results of transcriptional complex analysis and GATA3-ChIP-seq, ATAC-seq, and scRNA-seq trajectory analyses establish a positive regulatory role for the transcription factor GATA3 in IL-13 production by liver ILC2s, while AP-1 family members are shown to suppress IL-13 release. Thus, we identify a regulatory role and molecular mechanism by which liver ILC2s contribute to glucose homeostasis.
  • Jesus M Gomez-Salinero, Tomer Itkin, Chaitanya Badwe, Yang Lin, Sean Houghton, Balvir Kunar, Graeme M Birdsey, Victoria Kalna, Neil Dufton, Claire R Peghairx, Masataka Yokoyama, Matthew Wingo, Ge Li, Jenny Zhaoying Xiang, Yen-Michael Sheng Hsu, David Redmond, Ryan Schreiner, Anna M Randi, Shahin Rafii
    Nature Cardiovascular Research in Press 2022年7月  
  • Hiroaki Ochiiwa, Guzhanuer Ailiken, Masataka Yokoyama, Kazuyuki Yamagata, Hidekazu Nagano, Chihoko Yoshimura, Hiromi Muraoka, Keiji Ishida, Tomonori Haruma, Akitoshi Nakayama, Naoko Hashimoto, Kazutaka Murata, Motoi Nishimura, Yusuke Kawashima, Osamu Ohara, Shuichi Ohkubo, Tomoaki Tanaka
    Oncogene 40(7) 1217-1230 2021年2月  
    TAS4464, a potent, selective small molecule NEDD8-activating enzyme (NAE) inhibitor, leads to inactivation of cullin-RING E3 ubiquitin ligases (CRLs) and consequent accumulations of its substrate proteins. Here, we investigated the antitumor properties and action mechanism of TAS4464 in acute myeloid leukemia (AML). TAS4464 induced apoptotic cell death in various AML cell lines. TAS4464 treatments resulted in the activation of both the caspase-9-mediated intrinsic apoptotic pathway and caspase-8-mediated extrinsic apoptotic pathway in AML cells; combined treatment with inhibitors of these caspases markedly diminished TAS4464-induced apoptosis. In each apoptotic pathway, TAS4464 induced the mRNA transcription of the intrinsic proapoptotic factor NOXA and decreased that of the extrinsic antiapoptotic factor c-FLIP. RNA-sequencing analysis showed that the signaling pathway of the CRL substrate c-Myc was enriched after TAS4464 treatment. Chromatin immunoprecipitation (ChIP) assay revealed that TAS4464-induced c-Myc bound to the PMAIP1 (encoding NOXA) and CFLAR (encoding c-FLIP) promoter regions, and siRNA-mediated c-Myc knockdown neutralized both TAS4464-mediated NOXA induction and c-FLIP downregulation. TAS4464 activated both caspase-8 and caspase-9 along with an increase in NOXA and a decrease in c-FLIP, resulting in complete tumor remission in a human AML xenograft model. These findings suggest that NAE inhibition leads to anti-AML activity via a novel c-Myc-dependent apoptosis induction mechanism.
  • Brisa Palikuqi, Duc-Huy T. Nguyen, Ge Li, Ryan Schreiner, Alessandro F. Pellegata, Ying Liu, David Redmond, Fuqiang Geng, Yang Lin, Jesus M. Gómez-Salinero, Masataka Yokoyama, Paul Zumbo, Tuo Zhang, Balvir Kunar, Mavee Witherspoon, Teng Han, Alfonso M. Tedeschi, Federico Scottoni, Steven M. Lipkin, Lukas Dow, Olivier Elemento, Jenny Z. Xiang, Koji Shido, Jason R. Spence, Qiao J. Zhou, Robert E. Schwartz, Paolo De Coppi, Sina Y. Rabbany, Shahin Rafii
    Nature 585(7825) 426-432 2020年9月17日  
  • David M. Barry, Elizabeth A. McMillan, Balvir Kunar, Raphael Lis, Tuo Zhang, Tyler Lu, Edward Daniel, Masataka Yokoyama, Jesus M. Gomez-Salinero, Angara Sureshbabu, Ondine Cleaver, Annarita Di Lorenzo, Mary E. Choi, Jenny Xiang, David Redmond, Sina Y. Rabbany, Thangamani Muthukumar, Shahin Rafii
    Nature Communications 10(1) 2019年12月  
    Abstract Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders.
  • Masataka Yokoyama, Ippei Shimizu, Ayako Nagasawa, Yohko Yoshida, Goro Katsuumi, Takayuki Wakasugi, Yuka Hayashi, Ryutaro Ikegami, Masayoshi Suda, Yusuke Ota, Sho Okada, Marcus Fruttiger, Yoshio Kobayashi, Masanori Tsuchida, Yoshiaki Kubota, Tohru Minamino
    Journal of molecular and cellular cardiology 129 105-117 2019年4月  査読有り
    p53 is a guardian of the genome that protects against carcinogenesis. There is accumulating evidence that p53 is activated with aging. Such activation has been reported to contribute to various age-associated pathologies, but its role in vascular dysfunction is largely unknown. The aim of this study was to investigate whether activation of endothelial p53 has a pathological effect in relation to endothelial function. We established endothelial p53 loss-of-function and gain-of-function models by breeding endothelial-cell specific Cre mice with floxed Trp53 or floxed Mdm2/Mdm4 mice, respectively. Then we induced diabetes by injection of streptozotocin. In the diabetic state, endothelial p53 expression was markedly up-regulated and endothelium-dependent vasodilatation was significantly impaired. Impairment of vasodilatation was significantly ameliorated in endothelial p53 knockout (EC-p53 KO) mice, and deletion of endothelial p53 also significantly enhanced the induction of angiogenesis by ischemia. Conversely, activation of endothelial p53 by deleting Mdm2/Mdm4 reduced both endothelium-dependent vasodilatation and ischemia-induced angiogenesis. Introduction of p53 into human endothelial cells up-regulated the expression of phosphatase and tensin homolog (PTEN), thereby reducing phospho-eNOS levels. Consistent with these results, the beneficial impact of endothelial p53 deletion on endothelial function was attenuated in EC-p53 KO mice with an eNOS-deficient background. These results show that endothelial p53 negatively regulates endothelium-dependent vasodilatation and ischemia-induced angiogenesis, suggesting that inhibition of endothelial p53 could be a novel therapeutic target in patients with metabolic disorders.
  • Nagano H, Hashimoto N, Nakayama A, Suzuki S, Miyabayashi Y, Yamato A, Higuchi S, Fujimoto M, Sakuma I, Beppu M, Yokoyama M, Suzuki Y, Sugano S, Ikeda K, Tatsuno I, Manabe I, Yokote K, Inoue S, Tanaka T
    Proceedings of the National Academy of Sciences of the United States of America 115(33) 8370-8375 2018年8月  査読有り
  • Yokoyama M, Rafii S
    Nature cell biology 20(4) 365-366 2018年4月  査読有り
  • William Schachterle, Chaitanya R. Badwe, Brisa Palikuqi, Balvir Kunar, Michael Ginsberg, Raphael Lis, Masataka Yokoyama, Olivier Elemento, Joseph M. Scandura, Shahin Rafii
    NATURE COMMUNICATIONS 8 13963 2017年1月  査読有り
    Transplanting vascular endothelial cells (ECs) to support metabolism and express regenerative paracrine factors is a strategy to treat vasculopathies and to promote tissue regeneration. However, transplantation strategies have been challenging to develop, because ECs are difficult to culture and little is known about how to direct them to stably integrate into vasculature. Here we show that only amniotic cells could convert to cells that maintain EC gene expression. Even so, these converted cells perform sub-optimally in transplantation studies. Constitutive Akt signalling increases expression of EC morphogenesis genes, including Sox17, shifts the genomic targeting of Fli1 to favour nearby Sox consensus sites and enhances the vascular function of converted cells. Enforced expression of Sox17 increases expression of morphogenesis genes and promotes integration of transplanted converted cells into injured vessels. Thus, Ets transcription factors specify non-vascular, amniotic cells to EC-like cells, whereas Sox17 expression is required to confer EC function.
  • Takashi K. Ito, Masataka Yokoyama, Yohko Yoshida, Aika Nojima, Hidetoshi Kassai, Kengo Oishi, Sho Okada, Daisuke Kinoshita, Yoshio Kobayashi, Marcus Fruttiger, Atsu Aiba, Tohru Minamino
    PLOS ONE 9(7) e102186 2014年7月  査読有り
    Risk factors for atherosclerosis accelerate the senescence of vascular endothelial cells and promote atherogenesis by inducing vascular inflammation. A hallmark of endothelial senescence is the persistent up-regulation of pro-inflammatory genes. We identified CDC42 signaling as a mediator of chronic inflammation associated with endothelial senescence. Inhibition of CDC42 or NF-kappa B signaling attenuated the sustained up-regulation of pro-inflammatory genes in senescent human endothelial cells. Endothelium-specific activation of the p53/p21 pathway, a key mediator of senescence, also resulted in up-regulation of pro-inflammatory molecules in mice, which was reversed by Cdc42 deletion in endothelial cells. Likewise, endothelial-specific deletion of Cdc42 significantly attenuated chronic inflammation and plaque formation in atherosclerotic mice. While inhibition of NF-kappa B suppressed the pro-inflammatory responses in acute inflammation, the influence of Cdc42 deletion was less marked. Knockdown of cdc-42 significantly down-regulated pro-inflammatory gene expression and restored the shortened lifespan to normal in mutant worms with enhanced inflammation. These findings indicate that the CDC42 pathway is critically involved in senescence-associated inflammation and could be a therapeutic target for chronic inflammation in patients with age-related diseases without compromising host defenses.
  • Masataka Yokoyama, Sho Okada, Atsushi Nakagomi, Junji Moriya, Ippei Shimizu, Aika Nojima, Yohko Yoshida, Harumi Ichimiya, Naomi Kamimura, Yoshio Kobayashi, Shigeo Ohta, Marcus Fruttiger, Guillermina Lozano, Tohru Minamino
    CELL REPORTS 7(5) 1691-1703 2014年6月  査読有り
    Accumulating evidence has suggested a role for p53 activation in various age-associated conditions. Here, we identified a crucial role of endothelial p53 activation in the regulation of glucose homeostasis. Endothelial expression of p53 was markedly upregulated when mice were fed a high-calorie diet. Disruption of endothelial p53 activation improved dietary inactivation of endothelial nitric oxide synthase that upregulated the expression of peroxisome proliferator-activated receptor-gamma coactivator-1 alpha in skeletal muscle, thereby increasing mitochondrial biogenesis and oxygen consumption. Mice with endothelial cell-specific p53 deficiency fed a high-calorie diet showed improvement of insulin sensitivity and less fat accumulation, compared with control littermates. Conversely, upregulation of endothelial p53 caused metabolic abnormalities. These results indicate that inhibition of endothelial p53 could be a novel therapeutic target to block the vicious cycle of cardiovascular and metabolic abnormalities associated with obesity.
  • Sho Okada, Masataka Yokoyama, Haruhiro Toko, Kaoru Tateno, Junji Moriya, Ippei Shimizu, Aika Nojima, Takashi Ito, Yohko Yoshida, Yoshio Kobayashi, Hideki Katagiri, Tohru Minamino, Issei Komuro
    ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY 32(8) 1902-1909 2012年8月  査読有り
    Objective-The central nervous system is thought to influence the regulation of the cardiovascular system in response to humoral and neural signals from peripheral tissues, but our understanding of the molecular mechanisms involved is still quite limited. Methods and Results-Here, we demonstrate a central nervous system-mediated mechanism by which brain-derived neurotrophic factor (BDNF) has a protective effect against cardiac remodeling after myocardial infarction (MI). We generated conditional BDNF knockout mice, in which expression of BDNF was systemically reduced, by using the inducible Cre-loxP system. Two weeks after MI was induced surgically in these mice, systolic function was significantly impaired and cardiac size was markedly increased in conditional BDNF knockout mice compared with controls. Cardiomyocyte death was increased in these mice, along with decreased expression of survival molecules. Deletion of the BDNF receptor (tropomyosin-related kinase B) from the heart also led to the exacerbation of cardiac dysfunction after MI. The plasma levels of BDNF were markedly increased after MI, and this increase was associated with the upregulation of BDNF expression in the brain, but not in the heart. Ablation of afferent nerves from the heart or genetic disruption of neuronal BDNF expression inhibited the increase of plasma BDNF after MI and led to the exacerbation of cardiac dysfunction. Peripheral administration of BDNF significantly restored the cardiac phenotype of neuronal BDNF-deficient mice. Conclusion-These results suggest that BDNF expression is upregulated by neural signals from the heart after MI and then protects the myocardium against ischemic injury. (Arterioscler Thromb Vasc Biol 2012;32:1902-1909.)
  • Shimizu Ippei, Yoshida Yohko, Katsuno Taro, Tateno Kaoru, Okada Sho, Moriya Junji, Yokoyama Masataka, Nojima Aika, Ito Takashi, Zechner Rudolf, Komuro Issei, Kobayashi Yoshio, Minamino Tohru
    CELL METABOLISM 15(5) 787 2012年5月2日  査読有り
  • Ippei Shimizu, Yohko Yoshida, Taro Katsuno, Kaoru Tateno, Sho Okada, Junji Moriya, Masataka Yokoyama, Aika Nojima, Takashi Ito, Rudolf Zechner, Issei Komuro, Yoshio Kobayashi, Tohru Minamino
    CELL METABOLISM 15(1) 51-64 2012年1月  査読有り
    Several clinical studies have shown that insulin resistance is prevalent among patients with heart failure, but the underlying mechanisms have not been fully elucidated. Here, we report a mechanism of insulin resistance associated with heart failure that involves upregulation of p53 in adipose tissue. We found that pressure overload markedly upregulated p53 expression in adipose tissue along with an increase of adipose tissue inflammation. Chronic pressure overload accelerated lipolysis in adipose tissue. In the presence of pressure overload, inhibition of lipolysis by sympathetic denervation significantly downregulated adipose p53 expression and inflammation, thereby improving insulin resistance. Likewise, disruption of p53 activation in adipose tissue attenuated inflammation and improved insulin resistance but also ameliorated cardiac dysfunction induced by chronic pressure overload. These results indicate that chronic pressure overload upregulates adipose tissue p53 by promoting lipolysis via the sympathetic nervous system, leading to an inflammatory response of adipose tissue and insulin resistance.
  • Ippei Shimizu, Tohru Minamino, Haruhiro Toko, Sho Okada, Hiroyuki Ikeda, Noritaka Yasuda, Kaoru Tateno, Junji Moriya, Masataka Yokoyama, Aika Nojima, Gou Young Koh, Hiroshi Akazawa, Ichiro Shiojima, C. Ronald Kahn, E. Dale Abel, Issei Komuro
    JOURNAL OF CLINICAL INVESTIGATION 120(5) 1506-1514 2010年5月  査読有り
    Although many animal studies indicate insulin has cardioprotective effects, clinical studies suggest a link between insulin resistance (hyperinsulinemia) and heart failure (HF). Here we have demonstrated that excessive cardiac insulin signaling exacerbates systolic dysfunction induced by pressure overload in rodents. Chronic pressure overload induced hepatic insulin resistance and plasma insulin level elevation. In contrast, cardiac insulin signaling was upregulated by chronic pressure overload because of mechanical stretch-induced activation of cardiomyocyte insulin receptors and upregulation of insulin receptor and Irs1 expression. Chronic pressure overload increased the mismatch between cardiomyocyte size and vascularity, thereby inducing myocardial hypoxia and cardiomyocyte death. Inhibition of hyperinsulinemia substantially improved pressure overload-induced cardiac dysfunction, improving myocardial hypoxia and decreasing cardiomyocyte death. Likewise, the cardiomyocyte-specific reduction of insulin receptor expression prevented cardiac ischemia and hypertrophy and attenuated systolic dysfunction due to pressure overload. Conversely, treatment of type 1 diabetic mice with insulin improved hyperglycemia during pressure overload, but increased myocardial ischemia and cardiomyocyte death, thereby inducing HF. Promoting angiogenesis restored the cardiac dysfunction induced by insulin treatment. We therefore suggest that the use of insulin to control hyperglycemia could be harmful in the setting of pressure overload and that modulation of insulin signaling is crucial for the treatment of HF.
  • Moriya Junji, Minamino Tohru, Tateno Kaoru, Okada Sho, Shimizu Ippei, Nojima Aika, Yokoyama Masataka, Komuro Issei
    CIRCULATION 120(18) S1142 2009年11月3日  査読有り
  • Tohru Minamino, Masayuki Orimo, Ippei Shimizu, Takeshige Kunieda, Masataka Yokoyama, Takashi Ito, Aika Nojima, Akira Nabetani, Yuichi Oike, Hisahiro Matsubara, Fuyuki Ishikawa, Issei Komuro
    NATURE MEDICINE 15(9) 1082-U140 2009年9月  査読有り
    Various stimuli, such as telomere dysfunction and oxidative stress, can induce irreversible cell growth arrest, which is termed 'cellular senescence'(1,2). This response is controlled by tumor suppressor proteins such as p53 and pRb. There is also evidence that senescent cells promote changes related to aging or age-related diseases(3-6). Here we show that p53 expression in adipose tissue is crucially involved in the development of insulin resistance, which underlies age-related cardiovascular and metabolic disorders. We found that excessive calorie intake led to the accumulation of oxidative stress in the adipose tissue of mice with type 2 diabetes-like disease and promoted senescence-like changes, such as increased activity of senescence-associated beta-galactosidase, increased expression of p53 and increased production of proinflammatory cytokines. Inhibition of p53 activity in adipose tissue markedly ameliorated these senescence-like changes, decreased the expression of proinflammatory cytokines and improved insulin resistance in mice with type 2 diabetes-like disease. Conversely, upregulation of p53 in adipose tissue caused an inflammatory response that led to insulin resistance. Adipose tissue from individuals with diabetes also showed senescence-like features. Our results show a previously unappreciated role of adipose tissue p53 expression in the regulation of insulin resistance and suggest that cellular aging signals in adipose tissue could be a new target for the treatment of diabetes.
  • Yosuke Kayama, Tohru Minamino, Haruhiro Toko, Masaya Sakamoto, Ippei Shimizu, Hidehisa Takahashi, Sho Okada, Kaoru Tateno, Junji Moriya, Masataka Yokoyama, Aika Nojima, Michihiro Yoshimura, Kensuke Egashira, Hiroyuki Aburatani, Issei Komuro
    JOURNAL OF EXPERIMENTAL MEDICINE 206(7) 1565-1574 2009年7月  査読有り
    To identify a novel target for the treatment of heart failure, we examined gene expression in the failing heart. Among the genes analyzed, Alox15 encoding the protein 12/15 lipoxygenase (LOX) was markedly up-regulated in heart failure. To determine whether increased expression of 12/15-LOX causes heart failure, we established transgenic mice that overexpressed 12/15-LOX in cardiomyocytes. Echocardiography showed that Alox15 transgenic mice developed systolic dysfunction. Cardiac fibrosis increased in Alox15 transgenic mice with advancing age and was associated with the infiltration of macrophages. Consistent with these observations, cardiac expression of monocyte chemoattractant protein 1 (MCP-1) was up-regulated in Alox15 transgenic mice compared with wild-type mice. Treatment with 12-hydroxy-eicosatetraenoic acid, a major metabolite of 12/15-LOX, increased MCP-1 expression in cardiac fibroblasts and endothelial cells but not in cardiomyocytes. Inhibition of MCP-1 reduced the infiltration of macrophages into the myocardium and prevented both systolic dysfunction and cardiac fibrosis in Alox15 transgenic mice. Likewise, disruption of 12/15-LOX significantly reduced cardiac MCP-1 expression and macrophage infiltration, thereby improving systolic dysfunction induced by chronic pressure overload. Our results suggest that cardiac 12/15-LOX is involved in the development of heart failure and that inhibition of 12/15-LOX could be a novel treatment for this condition.

MISC

 55

講演・口頭発表等

 37

共同研究・競争的資金等の研究課題

 6